COPYRIGHTpsasir.upm.edu.my/id/eprint/76338/1/FPV 2018 24 IR.pdf · 2019. 12. 2. · meningkatkan...

58
UNIVERSITI PUTRA MALAYSIA COCKLE SHELL-DERIVED NANO CARRIER FOR ARA-C IN THE TREATMENT OF ACUTE MYELOID LEUKAEMIA MUSTAFA SADDAM GHAJI FPV 2018 24

Transcript of COPYRIGHTpsasir.upm.edu.my/id/eprint/76338/1/FPV 2018 24 IR.pdf · 2019. 12. 2. · meningkatkan...

  • © CO

    PYRI

    GHT U

    PM

    UNIVERSITI PUTRA MALAYSIA

    COCKLE SHELL-DERIVED NANO CARRIER FOR ARA-C IN THE TREATMENT OF ACUTE MYELOID LEUKAEMIA

    MUSTAFA SADDAM GHAJI

    FPV 2018 24

  • © CO

    PYRI

    GHT U

    PMCOCKLE SHELL-DERIVED NANO CARRIER FOR ARA-C IN THE

    TREATMENT OF ACUTE MYELOID LEUKAEMIA

    By

    MUSTAFA SADDAM GHAJI

    Thesis Submitted to the School of Graduate Studies, Universiti Putra Malaysia,

    in Fulfilment of the Requirements for the Degree of

    Doctor of Philosophy

    June 2018

  • © CO

    PYRI

    GHT U

    PM

    COPYRIGHT

    All material contained within the thesis, including without limitation text, logos, icons,

    photographs, and all other artwork, is copyright material of Universiti Putra Malaysia

    unless otherwise stated. Use may be made of any material contained within the thesis

    for non-commercial purposes from the copyright holder. Commercial use of material

    may only be made with the express, prior, written permission of Universiti Putra

    Malaysia.

    Copyright© Universiti Putra Malaysia

  • © CO

    PYRI

    GHT U

    PM

    DEDICATIONS

    To

    My Dear Parents,

    Mr. Saddam Ghaji

    Mrs. Sbeha Abod

    To

    My brothers and sisters

    To

    My Marvellous Family

    In Recognition of their Worth, Love, and Respect

  • © CO

    PYRI

    GHT U

    PM

    i

    Abstract of thesis presented to the senate of Universiti Putra Malaysia in fulfilment

    of the requirements for the degree of Doctor of Philosophy

    COCKLE SHELL-DERIVED NANO CARRIER FOR ARA-C IN THE

    TREATMENT OF ACUTE MYELOID LEUKAEMIA

    By

    MUSTAFA SADDAM GHAJI

    June 2018

    Chairman : Professor Md Zuki bin Abu Bakar, PhD

    Faculty : Veterinary Medicine

    Leukemia is a cancerous disease of bone marrow and blood in which acute form

    progresses more rapidly than the chronic form. The major therapeutic approaches of

    different cancer types are limited to conventional chemotherapy such as (Ara-C) which

    suffers less specific, high toxicity and short half-life, multidrug resistance and

    selectivity, narrow therapeutic index and significant increases in high dose distribution

    to healthy cells or tissues. Targeting anticancer drug delivery system has the potential

    to overcome these significant drawbacks by improving chemotherapy drug efficacy,

    specific tumor targeting, enhance accumulation in tumor tissues or cells and minimize

    the systemic toxicity. Nanoparticles as drug delivery system enable unique approaches

    to cancer treatment. Over the last two decades, a large number of nanoparticle delivery

    systems have been developed for cancer therapy including organic and inorganic

    materials. Cockle shells (Anadara granosa) are found to be a rich natural resource for

    calcium carbonate aragonite. In this study, the cockle shell-derived calcium carbonate

    aragonite nanoparticles (CCANPs) were used as a carrier for Cytarabine (Ara-C) as a

    unique approach for cancer treatment. Nanoparticles were spherical-shaped when

    CCANPs was synthesized using the combination of chemical and mechanical method.

    The morphology and compositions of the products were characterized by Field

    Emission Scanning Electron Microscope (FE-SEM), Transmission Electron

    Microscope (TEM), Energy Dispersive X-ray (EDX), X-Ray Diffraction (XRD),

    Fourier Transform Infra-Red (FT-IR) and zeta potential. The anti-leukemia drug (Ara-

    C) was loaded into CCANPs. The spectrophotometer was used with a wavelength UV-

    invisible, to estimate the amount of loading and release profile of Ara-C. The results

    showed that the drugs (Ara-C) could be efficiently loaded into the CCANPs, and

    furthermore, the fast and sustained release of Ara-C was observed from the

    nanocarriers at pH 4.8 and slow release at pH 7.4, which shows pH-dependent

    properties. The nanoparticles were used as a carrier against HL-60

    human leukemia cells (in vitro study) and for cancer therapy in a murine xenograft

    model (SCID mice) (in vivo study). The in vitro evaluation showed IC50 values upon

  • © CO

    PYRI

    GHT U

    PM

    ii

    72 hours of treatment with pure Ara-C was 5μg/mL, and Ara-C loaded CCANPs was

    2.5μg/mL. Apoptosis was demonstrated by Cell Counting Reagent (SF), Flow

    Cytometry (FCM), Methylene blue (MB) and Fluorescent Microscope (FM) where

    apparently cellular uptake of Ara-C/CCANPs through endocytosis indicating a dose

    and time-dependent response relationship. Morphological observations by SEM

    revealed microvilli disappearance, cell shrinkage, membrane blebbing and the

    formation of apoptotic bodies, which confirmed both Ara-C and half dose of Ara-

    C/CCANPs induced apoptosis of HL-60 cells. In brief, Ara-C loaded CCANPs are

    more effective than pure Ara-C to human leukemia (HL-60) cells. In vivo study

    revealed that CCANPs nanocarrier significantly enhances the effects of Ara-C on

    AML through blood smear, bone marrow smear and histopathological survey for vital

    organs (heart, liver, lung, spleen and kidney) for severe combined immunodeficient

    (SCID) mice. The pharmacokinetic study showing significant effect between pure

    Ara-C 50mg/kg group, 100mg/kg CCANPs loaded with 50mg/kg Ara-C and half dose

    of loaded drug (25/50 mg/kg), the rate of release of the drug in the plasma was slow

    in the two groups of the drug-loaded compared to the pure drug. The study revealed a

    new biodegradable, biocompatible, non-toxic to health and pH-sensitive, CCANPs

    with a feasible promising potential for targeted delivery carriers of antitumor drugs.

    The results established strong evidence that CCANPs has excellent properties that

    make it an ideal candidate for biological drug delivery systems.

  • © CO

    PYRI

    GHT U

    PM

    iii

    Abstrak tesis yang dikemukakan kepada Senat Universiti Putra Malaysia sebagai

    memenuhi keperluan untuk ijazah Doktor Falsafah

    PEMBAWA NANO BERASASKAN KULIT KERANG UNTUK ARA-C

    DALAM RAWATAN LEUKIMIA MYELOID AKUT

    Oleh

    MUSTAFA SADDAM GHAJI

    Jun 2018

    Pengerusi : Profesor Md Zuki bin Abu Bakar, PhD

    Fakulti : Perubatan Veterinar

    Leukemia adalah penyakit kanser sumsum tulang dan darah dimana bentuk akut

    merebak lebih pantas dari bentuk kronik. Pendekatan rawatan utama untuk jenis

    kanser yang berlainan adalah terhad kepada kemoterapi seperti (Ara-C) konvensional

    yang kurang spesifik, tinggi toksisiti dan berjangka hayat yang pendek, bersifat

    memilih dan rintangan drug pelbagai, indeks terapeutik yang sempit dan peningkatan

    pengedaran dos yang tinggi kepada sel atau tisu yang sihat. Sistem penyampaian drug

    antikanser yang disasarkan berpotensi untuk mengatasi permasalahan ini dengan

    meningkatkan keberkesanan drug kemoterapi, sasaran tumor yang spesifik,

    meningkatkan pengumpulan drug pada tisu atau sel tumor dan mengurangkan toksisiti

    sistemik. Nanopartikel sebagai sistem penyampaian drug memungkinkan pendekatan

    unik kepada rawatan kanser. Selama dua dekad yang lalu, sebahagian besar sistem

    penyampaian nanopartikel untuk terapi kanser telah dicipta termasuk penggunaan

    bahan organik dan bukan organik. Kulit kerang (Anadara granosa) merupakan sumber

    aragonit kalsium karbonat yang tinggi. Di dalam kajian ini, nanopartikel aragonit

    kalsium karbonat dari kulit kerang (CCANPs) digunakan sebagai pengangkut untuk

    Cytarabine (Ara-C) sebagai pendekatan unik rawatan kanser. Nanopartikel yang

    terhasil daripada gabungan kaedah mekanikal dan kimia adalah berbentuk sfera.

    Morfologi dan komposisi CCANPs telah dikenalpasti melalui Mikroskop

    Pengimbasan Pelepasan Medan (FE-SEM), Mikrsokop Transmisi Elektron (TEM), X-

    ray Penyerak Tenaga (EDX), Transformasi Fourier Spektroskopi Infra Merah (FTIR)

    dan Potensi Zeta. Drug anti-leukemia (Ara-C) telah dimuatkan ke dalam CCANPs.

    Spektrofotometer dengan gelombang bebas ultra-ungu telah digunakan untuk menilai

    jumlah muatan dan profil pelepasan Ara-C. Keputusan menunjukkan drug Ara-C

    boleh dimuatkan secara berkesan ke dalam CCANPs dan sebagai tambahan, pelepasan

    Ara-C daripada pengangkut nano adalah pantas dan berterusan pada pH 4.8 dan

    perlahan pada pH 7.4, di mana ini menunjukkan sifat kecenderungannya terhadap pH.

    Pengangkut nanotelah digunakan sebagai pengangkut melawan sel leukemia manusia

    HL-60 (kajian in-vitro) dan rawatan kanser menggunakan model xenograf mencit

  • © CO

    PYRI

    GHT U

    PM

    iv

    (CISD) (di dalam kajian in-vivo). Penilaian in-vitro menunjukkan nilai IC50 selepas

    rawatan selama 72 jam dengan Ara-C adalah 5 µg/ml dan Ara-C/CCANPs adalah 2.5

    µg/ml. Proses apoptosis telah ditunjukkan melalui reagen Pengiraan Sel (SF),

    Sitometer Aliran (FCM), Methylene Biru (MB) dan Mikroskopi Flouresens (FM) di

    mana telah jelas pengambilan selular Ara-C/CCANPs ialah melalui endositosis yang

    menunjukkan perhubungan respon dos dan masa. Pemerhatian morfologi melalui

    SEM menunjukkan kehilangan mikrovili, pengecutan sel, pembengkakan membran

    dan pembentukan jasad apoptosis yang mengesahkan kedua-dua Ara-C dan Ara-

    C/CCANPs separa dos berjaya menghasilkan apoptosis ke atas HL-60. Secara ringkas,

    ARA-C/CCANPs adalah lebih sitotoksik dari Ara-C ke atas sel leukemia HL-60.

    Kajian in-vivo menunjukkan pengangkut nano CCANPs berjaya meningkatkan

    (P

  • © CO

    PYRI

    GHT U

    PM

    v

    ACKNOWLEDGEMENTS

    First of all, I thank Allah the Almighty for the great grace. Despite all the difficulties

    that faced me during my study, but his great mercy has given me the strength and

    perseverance to complete this study. I praise and thank him a lot. I would like to

    express my deep gratitude and appreciations towards my respected supervisor Prof.

    Dr. Md Zuki Abu Bakar for his wonderful advice, thoughtful guidance and continuous

    support. He encouraged me through the difficulties and inspired me to move to a

    higher level of the learning experience. I would also like to give my heartfelt thanks

    and deepest appreciation and gratitude to my co-supervisors Dr. Intan Shameha Abdul

    Razak, Associate Professor Dr. Mohd Hezmee Mohd Noor and Associate Prof. Dr.

    Hazilawati Hj Hamzah for the continuous support of my PhD study and related

    research, for their patience, motivation, and immense knowledge. I would also like to

    express my gratitude to University Putra Malaysia for giving me the opportunity to

    complete this research. Without both the financial aids and grants, this thesis may yet

    still incomplete. A very special thanks all lecturers, technicians and staffs from

    Veterinary Medicine, UPM for their efforts to the successful of this work. I am so

    grateful to Iraqi Ministry of Higher Education and scientific research and Basrah

    University / Veterinary Medicine / Basrah, for their financial support and gave me the

    opportunity to continue my PhD. study. Last but not least, I would like to appreciate

    my beloved parents, my siblings, my dearest wife, my beloved kids and my friends for

    their unending support throughout this study. Thank you for the love, encouragement

    and prayers since the commencement of this study. I would not have made it this far

    without them. Thank you all.

  • © CO

    PYRI

    GHT U

    PM

  • © CO

    PYRI

    GHT U

    PM

    vii

    This thesis was submitted to the Senate of Universiti Putra Malaysia and has been

    accepted as fulfillment of the requirement for the degree of Doctor of Philosophy.

    The members of the Supervisory Committee were as follows:

    Md Zuki Abu Bakar, PhD

    Professor

    Faculty of Veterinary Medicine

    Universiti Putra Malaysia

    (Chairman)

    Intan Shameha Abdul Razak, PhD

    Senior Lecturer

    Faculty of Veterinary Medicine

    Universiti Putra Malaysia

    (Member)

    Mohd Hezmee Mohd Noor, PhD

    Associate Professor

    Faculty of Veterinary Medicine

    Universiti Putra Malaysia

    (Member)

    Hazilawati Hj Hamzah, PhD Associate Professor

    Faculty of Veterinary Medicine

    Universiti Putra Malaysia

    (Member)

    ____________________________

    ROBIAH BINTI YUNUS, PhD Professor and Dean

    School of Graduate Studies

    Universiti Putra Malaysia

    Date

  • © CO

    PYRI

    GHT U

    PM

  • © CO

    PYRI

    GHT U

    PM

  • © CO

    PYRI

    GHT U

    PM

    x

    TABLE OF CONTENTS

    Page

    ABSTRACT i

    ABSTRAK iii

    ACKNOWLEDGEMENTS v

    APPROVAL vi

    DECLARATION viii

    LIST OF TABLES xiv

    LIST OF FIGURES xv

    LIST OF ABBREVIATIONS xxi

    CHAPTER

    1 INTRODUCTION 1 1.1 General Background 1 1.2 Problem Statements 2 1.3 Hypothesis 2 1.4 Research Question 2 1.5 Objectives of the Study 3

    1.5.1 Main Objective 3 1.5.2 Specific Objectives 3

    2 LITERATURE REVIEW 4 2.1 Leukemia 4 2.2 Acute Myeloid Leukemia (AML) 4 2.3 Risk Factors in AML 5 2.4 Epidemiology of AML 5 2.5 Diagnosis of AML 6 2.6 Hematopoiesis 6 2.7 Classification 7

    2.7.1 The French-American-British (FAB) Classification of AML 8

    2.7.2 World Health Organization (WHO) Classification of

    AML 9 2.8 Current Therapies for Leukemia 9

    2.8.1 Biological Therapy 10 2.8.2 Targeted Therapy 10

    2.8.3 Radiation Therapy 10 2.8.4 Stem Cell Transplant 11 2.8.5 Chemotherapy Drugs 11

    2.8.5.1 Cytarabine 11 2.8.5.2 Dose of Cytarabine 13

    2.9 Nanoparticles (NPs) 14 2.10 Factors that can Affect on Treatment of Leukaemia 16

    2.10.1 Dose of Drug 16

  • © CO

    PYRI

    GHT U

    PM

    xi

    2.10.2 Size and Shape of Nanoparticles 16 2.10.3 Amount of Drug Loading 17 2.10.4 Drug Release 17 2.10.5 Surface Charge 17

    2.11 Calcium Overview Information 17 2.11.1 Calcium Carbonate 18 2.11.2 Type of Calcium Carbonate in Nature 18

    2.12 Search Strategy and Selection 19 2.13 Mouse Model 26

    2.13.1 Preclinical Mouse Models of Leukemia 26 2.14 Summary 27

    3 SYNTHESIS AND CHARACTERISATION OF COCKLE SHELL-DERIVED CaCO3 NANOPARTICLES AND

    CONTROLLED RELEASE PROFILE OF CYTARABINE 28 3.1 Introduction 28 3.2 Materials and Methods 29

    3.2.1 Preparation of Micron-Size Powder of Cockle Shell-Derived CaCO3 Aragonite (CCAMPs) 29

    3.2.2 Preparation of Cockle Shell-Derived CaCO3 Aragonite Nanoparticles (CCANPs) 29 3.2.2.1 Via Mechanical Method 29 3.2.2.2 Via Combination of Chemical and Mechanical

    Method 31

    3.2.3 Morphology, Crystallinity and Surface Properties of CCANPs 31

    3.2.4 Ara-C Loading and Encapsulation Efficiency 32 3.2.4.1 Ara-C Loading (Water Solvent) and

    Encapsulation Efficiency 32 3.2.4.1 Ara-C loading (Alcohol Solvent) and the

    Encapsulation Efficiency 32 3.2.5 In vitro Controlled Drug Release Study 32

    3.3 Results and Discussions 33 3.3.1 Synthesis of CCANPs 33

    3.3.2 TEM and FESEM Analysis 33 3.3.3 Determination of λ max of CCANPs 36

    3.3.4 Elemental Composition of the Synthesized CCAMPs and CCANPs 37

    3.3.5 Powder XRD Analysis 39 3.3.6 FT-IR Spectroscopy 39 3.3.7 Particle Size and Zeta Potential 43

    3.3.8 The Ara-C Loading (Water Solvent) and Encapsulation Efficiency 45

    3.3.9 The Ara-C Loading (Alcohol Solvent) and Encapsulation Efficiency 45

    3.3.10 In vitro Drug Release 48

    3.4 Conclusion 49

  • © CO

    PYRI

    GHT U

    PM

    xii

    4 In vitro EVALUATION OF Cytarabine-LOADED CCANPs FOR THE TREATMENT OF ACUTE MYELOID LEUKEMIA 50 4.1 Introduction 50 4.2 Materials and Methods 52

    4.2.1 Chemicals and Test Media 52 4.2.2 Cell Culture (HL-60) 52 4.2.3 Storage of HL-60 Cells Seed Stock 52 4.2.4 Cytotoxic Effects of Ara-C, Ara-C/CCANPs and

    CCANPs on HL-60 Cells 53 4.2.5 Cellular Uptake Assay 53 4.2.6 Flow Cytometric Analysis of Apoptosis and Necrosis 54 4.2.7 Morphological Changes in HL-60 Cells 54

    4.2.8 Statistical Analysis 54 4.3 Results and Discussion 54

    4.3.1 Cell Activity (Cell Counter Reagent (SF) Assay) 54 4.3.2 Evidence of Apoptosis by Flow Cytometry (FCM) 57 4.3.3 Evidence of Apoptosis by Fluorescence Microscopy 61 4.3.4 Evidence of Apoptosis by SEM 64 4.3.5 Anticancer Potential 65

    4.4 Conclusions 65

    5 In vivo RELEASE OF CYTARABINE-LOADED COCKLE SHELL-DERIVED CALCIUM CARBONATE ARAGONITE

    NANOPARTICLES (ARA-C/CCANPs) 66

    5.1 Introduction 66 5.2 Materials and Methods 67

    5.2.1 Calibration Curve of Ara-C 67 5.2.2 In Vitro Release of Ara-C 68 5.2.3 Stability Studies of Ara-C/CCANPs 68 5.2.4 In vivo Release of Ara-C 69

    5.2.4.1 Experimental Animals 69 5.2.4.2 Pharmacokinetic Study 69 5.2.4.3 HPLC Linearity 69 5.2.4.4 Preparation of Stock Solutions and Quality

    Control of Samples 70 5.3 Results and Discussion 70

    5.3.1 In vitro Release of Drug 70 5.3.2 Stability Studies of Ara-C/CCANPs 70 5.3.3 Linearity and Quantification and Limits of Detection 71 5.3.4 Pharmacokinetic Study 73

    5.4 Conclusion 76

  • © CO

    PYRI

    GHT U

    PM

    xiii

    6 EFFECTS OF CYTARABINE-LOADED CALCIUM CARBONATE ARAGONITE NANOPARTICLE ON ACUTE

    MYELOID LEUKEMIA In vivo: A NEW APPROACH TOWARDS

    INCREASING THE SPECIFICITY AND REDUCING THE

    TOXICITY OF ANTI-LEUKEMIA DRUG 77 6.1 Introduction 77 6.2 Materials and Methods 79

    6.2.1 HL-60 Cell Line 79 6.2.2 Animal Xenograft Model and Experimental Design 79 6.2.3 Histopathological Examination 82 6.2.4 Anesthesia of Mice 82 6.2.5 Blood Collection 82

    6.2.6 Wright Staining of Blood Smears 83 6.2.7 Haemato-Toxicity Evaluation 83 6.2.8 Serum Biochemistry Evaluation 83 6.2.9 Bone Marrow Smear 83

    6.3 Results and Discussion 84 6.3.1 Establishment and Characterization of

    Xenotransplantation 84 6.3.2 Body Weight of Mice 84 6.3.3 Weight of Organs 85 6.3.4 Histopathological Examination 86

    6.4 Hematological Parameters 103 6.4.1 Serum Biochemistry 109

    6.4.2 Bone Marrow Smear 112 6.5 Conclusion 115

    7 GENERAL DISCUSSION 116 7.1 Conclusion 118 7.2 Recommendations for Future Research 118

    REFERENCES 119 APPENDICES 148 BIODATA OF STUDENT 158

    LIST OF PUBLICATIONS 159

  • © CO

    PYRI

    GHT U

    PM

    xiv

    LIST OF TABLES

    Table Page

    2.1 The French-American-British (FAB) classification of AML 8

    2.2 The classification of AML based on the WHO 2008 criteria 9

    2.3 Data extracted from the previous studies on drug nanocarriers 21

    3.1 Elemental compositions of CCANPs. 38

    3.2 Elemental compositions of CCAMPs. 38

    3.3 Drug loading and encapsulation efficiency of Ara-C into CCANPs 45

    3.4 Drug loading and encapsulation efficiency of Ara-C into CCANPs 46

    4.1 Cell viability percentage of Ara-C and Ara-C/CCANPs on HL-60 cells

    determined by FS assay at 24, 48 and 72 h 57

    5.1 Stability evaluation of Ara-C-CCANPs at 4°C, room temperature (25°C)

    and 40°C 71

    5.2 Pharmacokinetic profile of Ara-C in BALB/c mice plasma 72

    5.3 Data shows drug release study for Ara-C in plasma mice. Amount of

    drug released was determined from the calibration curve. * represent

    significant difference at p˂0.05 73

    6.1 Total WBC, differential white blood cell, RBC and PCV in SCID mice

    for all experimental groups. The results presented are Mean ± SE (n=3) 111

  • © CO

    PYRI

    GHT U

    PM

    xv

    LIST OF FIGURES

    Figure Page

    2.1 A normal hematopoietic process. This figure depicts the normal

    differentiation of HSCs into the respective blood components; this is an

    essential leukemic process 7

    2.2 Cytarabine(4-amino-1-ß-D-arabinofuranosyl-2 (1H)-pyrimidinone).

    Cytosine normally combines with Ribose/Deoxyribose sugar, has a

    molecular weight of 243.22, and a molecular formula of C9H13N3O5 13

    2.3 : Preferred reporting items for systematic reviews and meta-analyses flow

    diagram: screening procedure of selected articles 20

    3.1 Diagrams show the jar with strips of aluminum bars and cross-section

    show barrier and glass balls with CCANPs 30

    3.2 Diagram shows the Roll Mill with jar on the cylindrical (horizontal

    position) 30

    3.3 TEM micrograph of CCAMPs magnification 100 nm (A) and FESEM

    micrograph of CCAMPs were sized at a rate of 1mm and magnification

    X50000 (B) 34

    3.4 FESEM micrograph of (A) CCANPs prepared via mechanical method

    and (B) Ara-C/CCANPs were sized at a rate of 30nm and magnification

    X2000000 34

    3.5 TEM micrograph of CCANPs prepared via a mechanical method and the

    particles were sized at a rate of 40 nm (A). Ara-C/CCANPs (B),

    magnification X50000 35

    3.6 FESEM micrographs of (A) CCANPs prepared via a combination

    method and (B) Ara-C/CCANPs were sized at a rate of 25nm and

    magnification X100000 35

    3.7 TEM micrographs of (A) CCANPs prepared via a combination method

    and (B) Ara-C/CCANPs were sized at a rate of 35nm and magnification

    X50000 36

    3.8 The wavelength of Ara-C in the UV spectrum 37

    3.9 EDX spectral of CCANPs (A) and CCAMPs (B) 38

    3.10 XRD pattern for (A) Ara-C, (B) CCAMPs, (C) Ara-C/CCANPs and (D)

    CCANPs 39

    3.11 FTIR spectra of Ara-C/CCANP (A), CCANPs (B) and Ara-C pure (C) 41

    3.12 FTIR spectra of CCMPs 42

    3.13 Particle size distributions (PSD) for CCANPs (A); Zeta potential of

    CCANPs before loaded (B) and after loaded (C) with Ara-C 44

  • © CO

    PYRI

    GHT U

    PM

    xvi

    3.14 FTIR spectra of CCANPs (40 mg), with different concentration of Ara-

    C (10, 20, 40 mg) 47

    3.15 Drug release profile for Ara-C, and Ara-C/CCANPs. Each point is the

    average of three replications and the vertical bars represent standard

    deviations 48

    4.1 Relative cell counts of HL-60 cells incubated at various concentrations

    for 24, 48 and 72hr. Values represent the mean ± standard error of the

    mean (n = 3); denotes statistically significant differences from control

    (*p < 0.05) 56

    4.2 Results of Flow cytometry showing a different distribution of cell

    cytopathology in HL-60 cells (control) and treated with Ara-C alone and

    ½ Ara-C/CCANPs at 24, 48, and 72 h. viable cells are red; early

    apoptosis is green; late apoptosis is blue 59

    4.3 Quantitative estimation (%) of cell cytopathology in HL-60 cells

    untreated and treated with Ara-C (5µM/mL) and Ara-C/CCANPs

    (2.5µM/mL) showing percentages of viable cells (VC), early apoptosis

    (EA), late apoptosis (LA) and necrosis (N) at 24, 48, and 72 h. Bars

    with a different number of asterisks are significantly different at p <

    0.05 60

    4.4 Fluorescent micrographs of acridine orange/propidium iodide double-

    stained HL-60 cells treated with (A) pure Ara-C, (B) ½ Ara-C/CCANPs

    and (C) Untreated cells without any morphological changes, (A and B)

    Cells treated for 72 hours show more orange-colored staining, indicating

    early apoptosis (yellow arrows); X100. 63

    4.5 Scanning electron micrograph of (A) untreated cell, (B) Ara-C 5µM/ml,

    (C) Ara-C 2.5µM/ml loaded into CCANPs 5µg/ml and (D) Ara-C

    5µg/ml loaded into CCANPs 10µg/ml treated groups. Treatment groups

    show the presence of blebbing signaling apoptosis 64

    5.1 Cytarabine structure and formula C9H13N3O5 67

    5.2 Calibration curve of Ara-C (Spectrophotometer correlation = 0.995) 68

    5.3 Stability study of Ara-C-CCANPs at 4 °C, 25 °C and 40 °C 71

    5.4 Calibration curve of Ara-C by HPLC (Correlation = 0.998) 72

    5.5 Data shows drug release study for Ara-C in plasma mice. The pure Ara-

    C levele in the plasma was higher (p

  • © CO

    PYRI

    GHT U

    PM

    xvii

    6.1 Diagram showing the method used in the induction of leukemia in SCID

    mice 80

    6.2 Summary of the experimental design 81

    6.3 Graft shows animals in the experimental groups loss their weight before

    treatment, and then regain their weight once the treatment begin except

    for the negative control group. The normal control group continued to

    gain weight. The values are mean ± SD of four measurements (n = 4) for

    five groups analyzed using test one way ANOVA 85

    6.4 Weight of organs of all groups experience at the end of the experimental

    period. Data are normalized to Negative control group and expressed as

    mean ± SE of (n=4). *p < 0.05 compared with normal control and all

    treatments 86

    6.5 Photomicrograph of the liver from NOD/SCID mice of normal control

    group showing normal structure. Central vein. H&E; X200 87

    6.6 Photomicrograph of the liver from NOD/SCID mice induced AML

    without treatment in negative control group at 53 days shows the area of

    metastatic tumor cells (MTC) (1), necrosis (black arrows) and liver cell

    degeneration (yellow arrows). H&E; X200 87

    6.7 Photomicrograph of the liver from NOD/SCID mice induced AML

    treated with Ara-C at 40 days of treatment shows sign of liver toxicity.

    Congestion (1) and hemorrhage (2). H&E; X200 88

    6.8 Photomicrograph of the liver from NOD/SCID mice induced AML

    treated with Ara-C/CCANPs (50mg/kg Ara-C-loaded CCANPs

    100mg/kg) at 40 days of treatment shows moderate changes with no

    evidence of MTC. Slight heamorrage (1); and necrosis (2). H&E, X200 88

    6.9 Photomicrograph of the liver from NOD/SCID mice induced AML

    treated with ½ dose of Ara-C/CCANPs at 40 days of treatment shows

    mild changes with no evidence of MTC. Normal sinusoidal (1);

    vacuolation (2). H&E, X200. 89

    6.10 Photomicrograph of the lung from NOD/SCID mice of normal control

    group showing normal alveoli and bronchioles (1). H&E, X100 90

    6.11 Photomicrograph of the lung from NOD/SCID mice induced AML

    without treatment in negative control group at 53 days shows the area of

    MTC (1), congestion (2) and alveolar wall degeneration (3). H&E, X100

    90

    6.12 Photomicrograph of the lung from NOD/SCID mice induced AML

    treated with Ara-C at 40 days of treatment shows the alveolar wall

    degeneration (1) and congestion (2) with no evidence of MTC. H&E,

    X100 91

  • © CO

    PYRI

    GHT U

    PM

    xviii

    6.13 Photomicrograph of the lung from NOD/SCID mice induced AML

    treated with Ara-C/CCANPs (50mg/kg Ara-C-loaded CCANPs

    100mg/kg) at 40 days of treatment shows the evidence of congestion

    with no area of MTC. H&E, X200 91

    6.14 Photomicrograph of the lung from NOD/SCID mice induced AML

    treated with ½ Ara-C/CCANPs (25mg/kg Ara-C-loaded CCANPs

    50mg/kg) at 40 days of treatment shows the evidence of congestion with

    no area of MTC. H&E, X200 92

    6.15 Photomicrograph of the kidney from NOD/SCID mice of normal control

    group shows normal structure of kidney. H&E, X400 93

    6.16 Photomicrograph of the Kidney from NOD/SCID mice induced AML

    without treatment in negative control group at 53 days shows area of

    MTC (1), tubule necrosis (2) and hemorrhage (3). H&E, X400 93

    6.17 Photomicrograph of the Kidney from NOD/SCID mice induced AML

    treated with Ara-C alone at 40 days of treatment shows epithelial cells

    degeneration (1), tubule vaculation (2) and hemorrhage (3). H&E, X400

    94

    6.18 Photomicrograph of the Kidney from NOD/SCID mice induced AML

    treated with Ara-C/CCANPs (50mg/kg Ara-C-loaded CCANPs

    100mg/kg) at 40 days of treatment shows tubule necrosis (1) and

    hemorrhage (2). H&E, X200. 94

    6.19 Photomicrograph of the Kidney from NOD/SCID mice induced AML

    treated with ½ Ara-C/CCANPs at 40 days of treatment show tubule

    necrosis (1) and hemorrhage (2). H&E, X100 95

    6.20 Photomicrograph of the heart tissue from NOD/SCID mice of normal

    control group showing normal heart wall structure. H&E X200 96

    6.21 Photomicrograph of the heart from NOD/SCID mice induced AML

    without treatment in negative control group at 53 days shows the area of

    MTCs (1), vacuolation (2) and myocardial degeneration (3). H&E, X200

    97

    6.22 Photomicrograph of the heart tissue from NOD/SCID mice induced

    AML treated with Ara-C at 40 days of treatment shows congestion (1)

    and myocardial degeneration (2) . H &E X200 97

    6.23 Photomicrograph of the heart tissue from NOD/SCID mice induced

    AML treated with Ara-C/CCANPs (50mg/kg Ara-C-loaded CCANPs

    100mg/kg) at 40 days of treatment shows hemorrhage (1) and vaculation

    (2). H&E; X200 98

    6.24 Photomicrograph of the heart tissue from NOD/SCID mice induced

    AML treated with ½ dose Ara-C/CCANPs at 40 days of treatment shows

    hemorrhage. H&E; X200 98

  • © CO

    PYRI

    GHT U

    PM

    xix

    6.25 Photomicrograph of the spleen from NOD/SCID mice of normal control

    group showing the normal structure. White pulp (1), red pulp (2) and

    central arteriole (3). H&E; X100 99

    6.26 Photomicrograph of the spleen from NOD/SCID mice induced AML

    without treatment in negative control group at 53 days shows the area of

    MTC (yellow arrows), borderline lesion was diagnosed as loss of cells

    in the marginal zone (1). H&E; X400 100

    6.27 Photomicrograph of the spleen from NOD/SCID mice induced AML

    treated with Ara-C at 40 days of treatment shows extra-medullary

    erythroid haemopoiesis. Note the aggregation of erythroid cells in the

    red pulp. H&E; X400 100

    6.28 Photomicrograph of the spleen from NOD/SCID mice induced AML

    treated with Ara-C/CCANPs (50mg/kg Ara-C-loaded CCANPs

    100mg/kg) at 40 days of treatment shows loss of cells in the marginal

    zone (black arrows). H&E; X200 101

    6.29 Photomicrograph of the spleen from NOD/SCID mice induced AML

    treated with ½ dose Ara-C/CCANPs at 40 days of treatment shows loss

    of cells in the marginal zone. H&E; X200 101

    6.30 Photomicrograph of the blood smear from NOD/SCID mice induced

    AML without treatment in negative control group at 14 days after cancer

    induction shows high number of leukocyte. Wright staining, X400 104

    6.31 Photomicrograph of the blood smear from NOD/SCID mice induced

    AML without treatment in negative control group at 53 days shows

    increase number of leukocyte. Wright staining, X400 104

    6.32 Graph shows the total number of RBC of mice at day 53. Data are

    normalized to negative control group and expressed as mean ± SE of

    (n=4). *p < 0.05 compared with negative control group 105

    6.33 Graph shows the total number of WBC of mice at day 53. Data are

    normalized to Negative control group and expressed as mean ± SE of

    (n=4). *p < 0.05 compared with normal control and all treatments 106

    6.34 Photomicrograph of the blood smear from NOD/SCID mice induced

    AML treated with ½ Ara-C/CCANPs at 53 days shows normal range of

    WBC. Wright staining, X400 106

    6.35 Photomicrograph of the blood smear from NOD/SCID mice induced

    AML treated with Ara-C/CCANPs (50mg/kg Ara-C-loaded CCANPs

    100mg/kg) at 53 days shows normal range of WBC. Wright staining,

    X400 107

    6.36 Photomicrograph of the blood smear from NOD/SCID mice of normal

    control group showing normal range of WBC. Wright staining, X400 107

    6.37 Photomicrograph of the blood smear from NOD/SCID mice induced

    AML treated with Ara-C at 53 days shows normal range of WBC.

    Wright staining, X400 108

  • © CO

    PYRI

    GHT U

    PM

    xx

    6.38 Graph shows of packed cell volume (PCV) of mice at day 53. Data are

    normalized to Negative control group and expressed as mean ± SE of

    (n=4). *p < 0.05 compared with negative control group 108

    6.39 Differential WBC across treatment groups at day 53. *p < 0.05 compared

    with another groups 109

    6.40 Graph shows the amount of urea in mice at day 53. Data are normalized

    to Negative control group and expressed as mean ± SE of independent

    trials. *p < 0.05 compared with normal control and all treatments 110

    6.41 Graph shows the total amount of Bilirubin in mice at day 53. Data are

    normalized to Negative control group and expressed as mean ± SE of

    independent trials. *p < 0.05 compared with normal control and all

    treatments 110

    6.42 Photomicrograph of the bone marrow of NOD/SCID mice (normal

    control group) Wright’s–Giemsa, X200 113

    6.43 Photomicrograph of the bone marrow of NOD/SCID mice induced AML

    treated with Ara-C at 40 days of treatment shows polymorphonuclear

    cell (1), fat tissue (yellow arrows), erythroblast (2) and lymphoblast (3).

    Wright’s–Giemsa; X200 113

    6.44 Photomicrograph of the bone marrow NOD/SCID mice induced AML

    treated with Ara-C/CCANPs (50mg/kg Ara-C-loaded CCANPs

    100mg/kg) at 40 days of treatment shows fat tissue (yellow arrows),

    erythroblast (1), and lymphoblast (2). Wright’s–Giemsa; X200 114

    6.45 Photomicrograph of the bone marrow NOD/SCID mice induced AML

    treated with ½ Ara-C/CCANPs at 40 days of treatment shows

    polymorphonuclear cell (1), fat tissue (yellow arrows), erythroblast (2)

    and lymphoblast (3). Wright’s–Giemsa; X200 114

  • © CO

    PYRI

    GHT U

    PM

    xxi

    LIST OF ABBREVIATIONS

    AML Acute myeloid leukemia

    ALL Acute lymphoid leukemia

    CML Chronic Myeloid Leukemia

    CLL Chronic Lymphoid Leukemia

    Ara-C Cytarabine

    EDX Equipped dispersive x-ray

    DL Drug loaded

    LE Loading efficiency

    FESEM Field emission scanning electron microscopy

    FTIR Fourier transformed infrared

    CCANPs Calcium Carbonate Aragonite nanoparticles

    Ara-C/CCANPs Cytarabine-loaded Calcium Carbonate nanoparticles

    NP Nanoparticle

    PBS Phosphate Buffer Saline

    SEM Scanning electron microscopy

    TEM Transmission electron microscopy

    XRD X-ray diffraction

    Nm Nano meter

    Mg/m2 Milligram per square meter

    mL Milliliter

    µ Micro meter

    Kg Kilogram

    nM Nano Molar

  • © CO

    PYRI

    GHT U

    PM

    xxii

    µg/mL Microgram per Milliliter

    h Hour (s)

    IC50 Lethal concentration, 50%

    SCID mice Severe combined immunodeficient mice

    CO2 Carbon dioxide

    AO Acridine Orange

    ATCC American Type Culture Collection

    FBS Fetal bovine serum

    Min Minute

    Rpm Revolution per minute

    PI Propidium iodide

    UV Ultraviolet

    % Percentage

    DNA Deoxyribonucleic acid

    HD High dose

    LD Low dosage

    BM Bone marrow

    PSD Particle size distributions

    MTD Maximally tolerated dose

    ID Intermediate dose

    MTCs Metastatic tumour cells

  • © CO

    PYRI

    GHT U

    PM

    1

    CHAPTER 1

    1 INTRODUCTION

    1.1 General Background

    Cancers develop from an uncontrolled growth of body cells and can develop from any

    part of the body. Any tissue from any part of the body can become cancerous provided

    the necessary factors needed for its propagation are in place (Folkman, 1996).

    Leukemia is a cancerous condition which primarily affects the blood and bone

    marrow. In the case of acute myeloid leukemia (AML), it develops from the bone

    marrow and in most cases, spread to the blood system. AML can also migrate from

    the bone marrow to the spleen, the central nervous system, the liver, the testicles, and

    the lymph nodes (Valent et al., 2007). The global rate of leukemia cases in 2012 was

    put at 4,000,000, resulting in 3,000,000 deaths. A large number of people in the USA

    (around 24,500, made up of 14,300 males and 10,200 females) were predicted to die

    from leukemia-related conditions in 2017 (Siegel et al., 2017). AML is the commonest

    form of adult leukemia, which despite the intimidating statistics regarding its

    associated mortality and morbidity, the treatment options is still unsuccessful in its

    management. Chemotherapy, which aims at killing the dividing cells over time to

    restore the normal blood count of normal cells, has been the only option for AML

    management; but, chemotherapy is a highly toxic procedure which lacks specificity.

    Nano-carriers are a new method of improving the specificity of chemotherapeutic

    agents and reducing their toxicity level. Based on this, cockle shell (Anadara

    granosa)-derived calcium carbonate aragonite nanoparticles (CCANPs) was produced

    and used as a drug carrier in this study. The size of nanoparticles (NPs) was within the

    transitional zone between the corresponding bulk materials and the individual atoms

    or molecules. This helps to alter the material’s physicochemical properties and present

    a chance to increase the uptake capability and interaction with biological tissues. In

    the living cells, the combination of these events can have an adverse impact

    biologically, which otherwise, would not have been possible with the same material

    in a larger form (Moore, 2006). Calcium and its derivatives are the most vital

    components of teeth and bone, in fact, the osseous tissues of bone are primarily

    composed of inorganic calcium-derived composite materials (Bandyopadhyay-Ghosh,

    2008). Calcium carbonate (CaCO3) is the commonest calcium derivative with the

    longest history of applications in various fields, such as in the plastics, paper, paint,

    food, inks, and pharmaceutical industries (Biradar et al., 2011). In the modern times,

    CaCO3 has attracted medical attention owing to its high applicability. It is a cost-

    effective, safe, biocompatible, resorptive, accessible, and osteoconductive material

    (Biradar et al., 2011). Owing to its pH sensitivity and relatively slow degradation, it

    can be used as an agent for the controlled release of active substances such as drugs to

    maintain their tolerable serum concentration and for targeted delivery over time (Qian

    et al., 2011). This study focused on the synthesis, characterization, and application of

    CCANPs as an agent for the in vivo and in vitro controlled release of Ara-C to targeted

    cancer tissues.

  • © CO

    PYRI

    GHT U

    PM

    2

    1.2 Problem Statements

    Acute leukemia is an aggressive form of cancer that requires immediate treatment.

    AML multimodal chemotherapy is used to re-establish and normalize blood and bone

    marrow cell numbers and morphology. Ara-C is a traditional chemotherapeutic agent

    for the management of all types of leukemia (Gökbuget et al., 2011). It is similar to

    most cancer chemotherapies which target the S-phase of cell division (healthy and

    cancer cells). The strategy requires a prolonged period of cell exposure to highly toxic

    concentrations of the agents for cancer treatment (Gökbuget et al., 2011; Hamada et

    al., 2002). However, the activity of Ara-C is decreased by its rapid deamination to the

    biologically-inactive metabolite, uracil (Hamada et al., 2002). This rapid deamination

    led to a search for effective formulations of Ara-C that cannot be deaminated, but still,

    exhibit better pharmacokinetic parameters and protection for Ara-C. The chemical

    therapies for AML are often limited in their use by high systemic toxicity and low

    specificity. Drug delivery through carrier systems is presumed to avoid their side

    effects through a controlled biodistribution. These carriers can contribute towards the

    control of leukemia metastasis. A new natural approach at nano-scale needs to be

    developed which ensures an efficient and enhanced drug delivery for AML treatment.

    1.3 Hypothesis

    i. CCANPs is a biocompatible and non-toxic to normal cells in normal pH.

    ii. CCANPs can be used as nano-carrier in the management of AML.

    iii. CCANPs reduce effective dose of Ara-C into half.

    iv. CCANPs loaded Ara-C has therapeutic effects on reduced metastasis HL-60

    human cells to other organs.

    1.4 Research Question

    i. What is the in vitro drug release profile and biocompatibility of CCANPs

    loaded Ara-C?

    ii. How safe is CCANPs loaded Ara-C on the biological system in vivo?

    iii. How effective CCANPs loaded Ara-C in the treatment Acute Myeloid

    leukaemia?

    iv. How CCANPs loaded Ara-C can reduce metastasis AML in other body

    organs?

    v. How CCANPs loaded Ara-C can reduce the dose of Ara-C?

  • © CO

    PYRI

    GHT U

    PM

    3

    1.5 Objectives of the Study

    1.5.1 Main Objective

    This study was conducted with the aim of investigating the effectiveness of CCANPs

    loaded Ara-C in the treatment of AML.

    1.5.2 Specific Objectives

    i. To synthesise and characterize CCANPs, and evaluate the in-vitro release profile of CCANPs loaded Ara-C.

    ii. To determine the CCANPs loaded Ara-C in the treatment HL-60 human cells in vitro.

    iii. To determine the pharmacokinetic of CCANPs loaded Ara-C in vivo. iv. To evaluate the effectiveness of CCANPs loaded Ara-C in the treatment of

    AML in SCID mice-induced AML in vivo.

  • © CO

    PYRI

    GHT U

    PM

    119

    8 REFERENCES

    Acharya, S., and Sahoo, S. K. (2011). PLGA nanoparticles containing various

    anticancer agents and tumour delivery by EPR effect. Advanced drug delivery

    reviews, 63 (3), 170-183.

    Akbarzadeh, A., Zarghami, N., Mikaeili, H., Asgari, D., Goganian, A. M., Khiabani,

    H. K., Samiei, M., and Davaran, S. (2012). Synthesis, characterization, and in

    vitro evaluation of novel polymer-coated magnetic nanoparticles for controlled

    delivery of doxorubicin. Nanotechnology, science and applications, 5, 13.

    Al-Qubaisi, M. S., Rasedee, A., Flaifel, M. H., Ahmad, S. H., Hussein-Al-Ali, S.,

    Hussein, M. Z., Zainal, Z., Alhassan, F. H., Taufiq-Yap, Y. H., and Eid, E. E.

    (2013). Induction of apoptosis in cancer cells by NiZn ferrite nanoparticles

    through mitochondrial cytochrome C release. International Journal of

    Nanomedicine, 8, 4115.

    Alex, R., and Bodmeier, R. (1990). Encapsulation of water-soluble drugs by a

    modified solvent evaporation method. I. Effect of process and formulation

    variables on drug entrapment. Journal of Microencapsulation, 7 (3), 347-355.

    Alexis, F., Pridgen, E., Molnar, L. K., and Farokhzad, O. C. (2008). Factors affecting

    the clearance and biodistribution of polymeric nanoparticles. Molecular

    pharmaceutics, 5 (4), 505-515.

    Ambade, A. V., Savariar, E. N., and Thayumanavan, S. (2005). Dendrimeric micelles

    for controlled drug release and targeted delivery. Molecular pharmaceutics, 2

    (4), 264-272.

    Amin, M. R., Postma, G. N., Johnson, P., Digges, N., and Koufman, J. A. (2001).

    Proton pump inhibitor resistance in the treatment of laryngopharyngeal reflux.

    Otolaryngology—Head and Neck Surgery, 125 (4), 374-378.

    Anandhakumar, S., Nagaraja, V., and Raichur, A. M. (2010). Reversible

    polyelectrolyte capsules as carriers for protein delivery. Colloids and Surfaces

    B: Biointerfaces, 78 (2), 266-274.

    Appelbaum, F. R., Gundacker, H., Head, D. R., Slovak, M. L., Willman, C. L.,

    Godwin, J. E., Anderson, J. E., and Petersdorf, S. H. (2006). Age and acute

    myeloid leukemia. Blood, 107 (9), 3481-3485.

    Arnoux, P., and Morrison, R. (1992). Drug analysis of biological samples. A survey

    of validation approaches in chromatography in the UK pharmaceutical

    industry. Xenobiotica, 22 (7), 757-764.

    Aryal, S., Hu, C.-M. J., Fang, R. H., Dehaini, D., Carpenter, C., Zhang, D.-E., and

    Zhang, L. (2013). Erythrocyte membrane-cloaked polymeric nanoparticles for

    controlled drug loading and release. Nanomedicine, 8 (8), 1271-1280.

  • © CO

    PYRI

    GHT U

    PM

    120

    Aziz, N. B. A., and Al-Jubori, S. S. (2016). Genetic elements responsible for extreme

    drug resistance (Xdr) in Klebsiella Pnumoniae Var Pnumoniae isolated from

    clinical samples of Iraqi patients. World Journal Of Pharmaceutical Research,

    5 (5), 1-23.

    Bader, J. P. (1965). The requirement for DNA synthesis in the growth of Rous sarcoma

    and Rous-associated viruses. Virology, 26 (2), 253-261.

    Baguley, B., and Falkenhaug, E. (1971). Plasma half-life of cytosine arabinoside

    (NSC-63878) in patients treated for acute myeloblastic leukemia. Cancer

    chemotherapy reports, 55 (3), 291.

    Bandyopadhyay-Ghosh, S. (2008). Bone as a collagen-hydroxyapatite composite and

    its repair. Trends Biomater Artif Organs, 22 (2), 116-124.

    Banerjee, T., Mitra, S., Singh, A. K., Sharma, R. K., and Maitra, A. (2002).

    Preparation, characterization and biodistribution of ultrafine chitosan

    nanoparticles. International journal of pharmaceutics, 243 (1-2), 93-105.

    Bellone, M., Calcinotto, A., Filipazzi, P., De Milito, A., Fais, S., and Rivoltini, L.

    (2013). The acidity of the tumor microenvironment is a mechanism of immune

    escape that can be overcome by proton pump inhibitors. Oncoimmunology, 2

    (1), e22058.

    Bennett, J., Catovsky, D., Daniel, M., Flandrin, G., Galton, D., Gralnick, H., and

    Sultan, C. (1989). Proposals for the classification of chronic (mature) B and T

    lymphoid leukaemias. French-American-British (FAB) Cooperative Group.

    Journal of clinical pathology, 42 (6), 567-584.

    Bennett, J. M., Catovsky, D., Daniel, M. T., Flandrin, G., Galton, D. A., Gralnick, H.

    R., and Sultan, C. (1976). Proposals for the Classification of the Acute

    Leukaemias French‐American‐British (FAB) Co‐operative Group. British journal of haematology, 33 (4), 451-458.

    Bergmann, W., and Feeney, R. J. (1951). Contributions to the study of marine

    products. xxxii. The nucleosides of sponges. I. The Journal of Organic

    Chemistry, 16 (6), 981-987.

    Biradar, S., Ravichandran, P., Gopikrishnan, R., Goornavar, V., Hall, J. C., Ramesh,

    V., Baluchamy, S., Jeffers, R. B., and Ramesh, G. T. (2011). Calcium

    carbonate nanoparticles: synthesis, characterization and biocompatibility.

    Journal of nanoscience and nanotechnology, 11 (8), 6868-6874.

    Blanco, M., Trigo, R., Teijón, C., Gómez, C., and Teijón, J. (1998). Slow releasing of

    ara-C from poly (2-hydroxyethyl methacrylate) and poly (2-hydroxyethyl

    methacrylate-co-N-vinyl-2-pyrrolidone) hydrogels implanted subcutaneously

    in the back of rats. Biomaterials, 19 (7-9), 861-869.

  • © CO

    PYRI

    GHT U

    PM

    121

    Bonnet, D., and Dick, J. E. (1997). Human acute myeloid leukemia is organized as a

    hierarchy that originates from a primitive hematopoietic cell. Nature medicine,

    3 (7), 730-737.

    Borsa, J., Whitmore, G., Valeriote, F., Collins, D., and Bruce, W. (1969). Studies on

    the Persistence of Methotrexate, Cytosine Arabinoside, and Leucovorin in

    Serum of Mice 2. Journal of the National Cancer Institute, 42 (2), 235-242.

    Bosma, M. J., and Carroll, A. M. (1991). The SCID mouse mutant: definition,

    characterization, and potential uses. Annual Review of Immunology, 9 (1), 323-

    350.

    Brognard, J., Clark, A. S., Ni, Y., and Dennis, P. A. (2001). Akt/protein kinase B is

    constitutively active in non-small cell lung cancer cells and promotes cellular

    survival and resistance to chemotherapy and radiation. Cancer Res, 61 (10),

    3986-3997.

    Bryant, A. L., Deal, A. M., Walton, A., Wood, W. A., Muss, H., and Mayer, D. K.

    (2015). Use of ED and hospital services for patients with acute leukemia after

    induction therapy: one year follow-up. Leukemia Research, 39 (4), 406-410.

    Bunce, C., Fisher, A., Toksoz, D., and Brown, G. (1983). Isolation and

    characterisation of dimethylsulphoxide resistant variants from the human

    promyeloid cell line HL60. Experimental Hematology, 11 (9), 828-833.

    Bustamante, A., Giralt, D., Garcia‐Bonilla, L., Campos, M., Rosell, A., and Montaner, J. (2012). Citicoline in pre‐clinical animal models of stroke: a meta‐analysis shows the optimal neuroprotective profile and the missing steps for jumping

    into a stroke clinical trial. Journal of neurochemistry, 123 (2), 217-225.

    Calcinotto, A., Filipazzi, P., Grioni, M., Iero, M., De Milito, A., Ricupito, A., Cova,

    A., Canese, R., Jachetti, E., and Rossetti, M. (2012). Modulation of

    microenvironment acidity reverses anergy in human and murine tumor-

    infiltrating T lymphocytes. Cancer Res, 72 (11), 2746-2756.

    Caldas, S. S., Demoliner, A., and Primel, E. G. (2009). Validation of a method using

    solid phase extraction and liquid chromatography for the determination of

    pesticide residues in groundwaters. Journal of the Brazilian Chemical Society,

    20 (1), 125-132.

    Capizzi, R. L., Yang, J.-L., Cheng, E., Bjornsson, T., Sahasrabudhe, D., Tan, R., and

    Cheng, Y. (1983). Alteration of the pharmacokinetics of high-dose ara-C by

    its metabolite, high ara-U in patients with acute leukemia. Journal of Clinical

    Oncology, 1 (12), 763-771.

    Casanova, H., and Higuita, L. P. (2011). Synthesis of calcium carbonate nanoparticles

    by reactive precipitation using a high pressure jet homogenizer. Chemical

    Engineering Journal, 175, 569-578.

  • © CO

    PYRI

    GHT U

    PM

    122

    Ch'ien, L. T., Schabel Jr, F. M., and Alford Jr, C. A. (1973). Arabinosyl nucleosides

    and nucleotides. Selective inhibitors of viral functions. CRC Press, Cleveland,

    227-256.

    Chakraborty, C., Pal, S., Doss, G., Wen, Z.H., and Lin, C.S. (2013). Nanoparticles as'

    smart'pharmaceutical delivery. Frontiers in bioscience (Landmark edition),

    18, 1030-1050.

    Chandran, P., Gupta, N., Retnakumari, A. P., Malarvizhi, G. L., Keechilat, P., Nair,

    S., and Koyakutty, M. (2013). Simultaneous inhibition of aberrant cancer

    kinome using rationally designed polymer-protein core-shell nanomedicine.

    Nanomedicine: Nanotechnology, Biology and Medicine, 9 (8), 1317-1327.

    Chandran, P., Kavalakatt, A., Malarvizhi, G. L., Vasanthakumari, D. R. V. N.,

    Retnakumari, A. P., Sidharthan, N., Pavithran, K., Nair, S., and Koyakutty, M.

    (2014). Epigenetics targeted protein-vorinostat nanomedicine inducing

    apoptosis in heterogeneous population of primary acute myeloid leukemia

    cells including refractory and relapsed cases. Nanomedicine: Nanotechnology,

    Biology and Medicine, 10 (4), 721-732.

    Chandran, S., and Singh, R. (2007). Comparison of various international guidelines

    for analytical method validation. Die Pharmazie-An International Journal of

    Pharmaceutical Sciences, 62 (1), 4-14.

    Chandy, T., Das, G. S., Wilson, R. F., and Rao, G. H. (2002). Development of

    polylactide microspheres for protein encapsulation and delivery. Journal of

    applied polymer science, 86 (5), 1285-1295.

    Cheng, R., Feng, F., Meng, F., Deng, C., Feijen, J., and Zhong, Z. (2011). Glutathione-

    responsive nano-vehicles as a promising platform for targeted intracellular

    drug and gene delivery. Journal of Controlled Release, 152 (1), 2-12.

    Clark, W. M. (2009). Efficacy of citicoline as an acute stroke treatment. Expert

    opinion on pharmacotherapy, 10 (5), 839-846.

    Clarke, M. L., Mackey, J. R., Baldwin, S. A., Young, J. D., and Cass, C. E. (2002).

    The role of membrane transporters in cellular resistance to anticancer

    nucleoside drugs. Clinically Relevant Resistance in Cancer Chemotherapy, 27-

    47.

    Cohen, S. S. (1966). Introduction to the biochemistry of D-arabinosyl nucleosides.

    Progress in nucleic acid research and molecular biology, 5, 1-88.

    Cohen, S. S., Flaks, J. G., Barner, H. D., Loeb, M. R., and Lichtenstein, J. (1958). The

    mode of action of 5-fluorouracil and its derivatives. Proceedings of the

    National Academy of Sciences, 44 (10), 1004-1012.

    Cohen, S. S., and Plunkett, W. (1975). The utilization of nucleotides by animal cells.

    Annals of the New York Academy of Sciences, 255 (1), 269-286.

  • © CO

    PYRI

    GHT U

    PM

    123

    Collins, S. J., Ruscetti, F. W., Gallagher, R. E., and Gallo, R. C. (1978). Terminal

    differentiation of human promyelocytic leukemia cells induced by dimethyl

    sulfoxide and other polar compounds. Proceedings of the National Academy

    of Sciences, 75 (5), 2458-2462.

    Cooper, J., and Hunt, J. (2006). The significance of zeta potential in osteogenesis.

    Paper presented at the Annual Meeting-Society for Biomaterials in

    Conjunction with the International Biomaterials Symposium.

    Cox, J. D., Stetz, J., and Pajak, T. F. (1995). Toxicity criteria of the radiation therapy

    oncology group (RTOG) and the European organization for research and

    treatment of cancer (EORTC). International Journal of Radiation Oncology

    Biology Physics, 31 (5), 1341-1346.

    Creasey, W. (1963). Studies on the Metabolism of 5-Iodo-2'-deoxycytidine in vitro

    purification of nucleoside deaminase from mouse kidney. Journal of

    Biological Chemistry, 238 (5), 1772-1776.

    Dash, S. K., Chattopadhyay, S., Dash, S. S., Tripathy, S., Das, B., Mahapatra, S. K.,

    Bag, B. G., Karmakar, P., and Roy, S. (2015). Self assembled nano fibers of

    betulinic acid: A selective inducer for ROS/TNF-alpha pathway mediated

    leukemic cell death. Bioorganic chemistry, 63, 85-100.

    Davis, M. E. (2009). The first targeted delivery of siRNA in humans via a self-

    assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic.

    Molecular pharmaceutics, 6 (3), 659-668.

    De Campos, A. M., Sánchez, A., and Alonso, M. a. J. (2001). Chitosan nanoparticles:

    a new vehicle for the improvement of the delivery of drugs to the ocular

    surface. Application to cyclosporin A. International journal of pharmaceutics,

    224 (1-2), 159-168.

    Dharmalingam, S. R., Ramamurthy, S., Chidambaram, K., and Nadaraju, S. (2014). A

    simple HPLC bioanalytical method for the determination of doxorubicin

    hydrochloride in rat plasma: application to pharmacokinetic studies. Tropical

    Journal of Pharmaceutical Research, 13 (3), 409-415.

    Dizaj, S. M., Lotfipour, F., Barzegar-Jalali, M., Zarrintan, M.-H., and Adibkia, K.

    (2016). Physicochemical characterization and antimicrobial evaluation of

    gentamicin-loaded CaCO3 nanoparticles prepared via microemulsion method.

    Journal of Drug Delivery Science and Technology, 35, 16-23.

    Doering, A., Keller, J., and Cohen, S. S. (1966). Some effects of D-arabinosyl

    nucleosides on polymer syntheses in mouse fibroblasts. Cancer Res, 26 (12

    Part 1), 2444-2450.

  • © CO

    PYRI

    GHT U

    PM

    124

    Dong, Q., Li, J., Cui, L., Jian, H., Wang, A., and Bai, S. (2017). Using porous

    CaCO3/hyaluronic acid nanocages to accommodate hydrophobic

    photosensitizer in aqueous media for photodynamic therapy. Colloids and

    Surfaces A: Physicochemical and Engineering Aspects, 516, 190-198.

    Doss, C. G. P., Debottam, S., and Debajyoti, C. (2013). Glutathione-responsive nano-

    transporter-mediated siRNA delivery: silencing the mRNA expression of Ras.

    Protoplasma, 250 (3), 787-792.

    Druker, B. J. (2002). STI571 (Gleevec™) as a paradigm for cancer therapy. Trends in

    molecular medicine, 8 (4), S14-S18.

    Duzgunes, N. (2012). Nanomedicine: infectious diseases, immunotherapy,

    diagnostics, antifibrotics, toxicology and gene medicine (Vol. 509), 1st edition:

    Elsevier Academic Press, USA.

    Early, A. P., Preisler, H. D., Slocum, H., and Rustum, Y. M. (1982). A pilot study of

    high-dose 1-β-D-arabinofuranosylcytosine for acute leukemia and refractory

    lymphoma: clinical response and pharmacology. Cancer Res, 42 (4), 1587-

    1594.

    El-Subbagh, H. I., and Al-Badr, A. A. (2009). Cytarabine Profiles of Drug Substances,

    Excipients and Related Methodology (Vol. 34, pp. 37-113): Elsevier Academic

    Press, USA.

    Emerich, D. F., and Thanos, C. G. (2006). The pinpoint promise of nanoparticle-based

    drug delivery and molecular diagnosis. Biomolecular engineering, 23 (4), 171-

    184.

    Estey, E. H. (2012). Acute myeloid leukemia: 2012 update on diagnosis, risk

    stratification, and management. American journal of hematology, 87 (1), 89-

    99.

    Evans, J. S., Musser, E. A., Bostwick, L., and Mengel, G. D. (1964). The effect of 1-

    β-D-arabinofuranosylcytosine hydrochloride on murine neoplasms. Cancer

    Res, 24 (7), 1285-1293.

    Evans, J. S., Musser, E. A., Mengel, G. D., Forsblad, K. R., and Hunter, J. H. (1961).

    Antitumor activity of 1-β-D-arabinofuranosylcytosine hydrochloride.

    Proceedings of the Society for Experimental Biology and Medicine, 106 (2),

    350-353.

    Ezat, W., Noraziani, K., and Sabrizan, O. (2012). Improving quality of life among

    cancer patients in Malaysia. Asian Pacific Journal of Cancer Prevention, 13

    (3), 1069-1075.

    Falini, B., and Tiacci, E. (2010). Hairy cell leukemia. The Lymphoid Neoplasms 3ed,

    471.

  • © CO

    PYRI

    GHT U

    PM

    125

    Falini, B., Tiacci, E., Martelli, M. P., Ascani, S., and Pileri, S. A. (2010). New

    classification of acute myeloid leukemia and precursor-related neoplasms:

    changes and unsolved issues. Discovery medicine, 10 (53), 281-292.

    Farokhzad, O. C., and Langer, R. (2006). Nanomedicine: developing smarter

    therapeutic and diagnostic modalities. Advanced Drug Delivery Reviews, 58

    (14), 1456-1459.

    Fernandez-Montesinos, R., Castillo, P. M., Klippstein, R., Gonzalez-Rey, E., Mejias,

    J. A., Zaderenko, A. P., and Pozo, D. (2009). Chemical synthesis and

    characterization of silver-protected vasoactive intestinal peptide nanoparticles.

    Nanomedicine, 4 (8), 919-930.

    Fiegl, M. (2016). Epidemiology, pathogenesis, and etiology of acute leukemia

    Handbook of Acute Leukemia (pp. 3-13): Springer.

    Field, J. B., Boryczka, A., and Costa, F. (1955). Tests of compounds against sarcoma

    180 in a screening project in mice. Cancer Res, 15 (Supplement 2), 3-52.

    Fierro, I. M., de Menezes Alencar, M. S., Lins Mendes, F. M., de Souza Mendes, C.

    d. U., Nunes, B. F., and de Souza Antunes, A. M. (2014). Nanoparticles applied

    to antineoplastic agents: a patent landscape. Pharmaceutical patent analyst, 3

    (6), 613-623.

    Flowers, C. R., and Armitage, J. O. (2010). A decade of progress in lymphoma:

    advances and continuing challenges. Clinical Lymphoma Myeloma and

    Leukemia, 10 (6), 414-423.

    Folkman, J. (1996). Fighting cancer by attacking its blood supply. Scientific American,

    275 (3), 150-154.

    Folley, J. H., Borges, W., and Yamawaki, T. (1952). Incidence of leukemia in

    survivors of the atomic bomb in Hiroshima and Nagasaki, Japan. The American

    journal of medicine, 13 (3), 311-321.

    Foon, K. A. (1986). Immunologic classification of leukemia and lymphoma. Blood,

    68 (1), 1-31.

    Foucar, K., Reichard, K., and Czuchlewski, D. (2010). Acute Myeloid Leukemia.

    Bone marrow pathology, 3rd edition. ASCP Press, Chicago, 377-431.

    Fox, J. J., Yung, N., and Bendich, A. (1957). Pyrimidine nucleosides. II. The synthesis

    of 1-β-D-arabinofuranosylthymine (“Spongothymidine”). Journal of the

    American Chemical Society, 79 (11), 2775-2778.

    Foye, W. O. (2008). Foye's principles of medicinal chemistry, 6th edition: Lippincott

    Williams & Wilkins, USA.

  • © CO

    PYRI

    GHT U

    PM

    126

    Frattini, M. G., and Maslak, P. G. (2008). Strategy for incorporating molecular and

    cytogenetic markers into acute myeloid leukemia therapy. Journal of the

    National Comprehensive Cancer Network, 6 (10), 995-1002.

    Freireich, E. J., Gehan, E., Rall, D., Schmidt, L., and Skipper, H. (1966). Quantitative

    comparison of toxicity of anticancer agents in mouse, rat, hamster, dog,

    monkey, and man. Cancer chemotherapy reports, 50 (4), 219.

    Frese, K. K., and Tuveson, D. A. (2007). Maximizing mouse cancer models. Nature

    Reviews Cancer, 7 (9), 654-658.

    Fröhling, S., Schlenk, R. F., Kayser, S., Morhardt, M., Benner, A., Döhner, K., and

    Döhner, H. (2006). Cytogenetics and age are major determinants of outcome

    in intensively treated acute myeloid leukemia patients older than 60 years:

    results from AMLSG trial AML HD98-B. Blood, 108 (10), 3280-3288.

    Fu, W., Mohd Noor, M. H., Yusof, L. M., Ibrahim, T. A. T., Keong, Y. S., Jaji, A. Z.,

    and Zakaria, M. Z. A. B. (2017). In vitro evaluation of a novel pH sensitive

    drug delivery system based cockle shell-derived aragonite nanoparticles

    against osteosarcoma. Journal of Experimental Nanoscience, 12 (1), 166-187.

    Fuller, B. (1987). Low temperature preservation in medicine and veterinary science.

    Effects of low temperatures on biological systems/edited by BWW Grout, GJ

    Morris

    Furth, J., and Cohen, S. S. (1967). Inhibition of mammalian DNA polymerase by the

    5′-triphosphate of 9-β-D-arabinofuranosyladenine. Cancer Res, 27 (9), 1528-

    1533.

    Furth, J., and Cohen, S. S. (1968). Inhibition of mammalian DNA polymerase by the

    5′-triphosphate of 1-β-D-arabinofuranosylcytosine and the 5′-triphosphate of

    9-β-D-arabinofuranosyladenine. Cancer Res, 28 (10), 2061-2067.

    Gamble, G. L., Gerber, L. H., Spill, G. R., and Paul, K. L. (2011). The future of cancer

    rehabilitation: emerging subspecialty. American journal of physical medicine

    & rehabilitation, 90 (5), S76-S87.

    Gentry, G. A., and Aswell, J. F. (1975). Inhibition of herpes simplex virus replication

    by araT. Virology, 65 (1), 294-296.

    Gerard, E. L., Ferry, J. A., Amrein, P. C., Harmon, D. C., Mckinstry, R. C., Hoppel,

    B. E., and Rosen, B. (1992). Compositional changes in vertebral bone marrow

    during treatment for acute leukemia: assessment with quantitative chemical

    shift imaging. Radiology, 183 (1), 39-46.

    Ghafar, S. L. M. A., Hussein, M. Z., Rukayadi, Y., and Zakaria, M. Z. A. B. (2017).

    Surface-functionalized cockle shell–based calcium carbonate aragonite

    polymorph as a drug nanocarrier. Nanotechnology, science and applications,

    10, 79.

  • © CO

    PYRI

    GHT U

    PM

    127

    Ghaji, M., Zakaria, Z. A. B., Shameha, A., Noor, M. H. M., and Hazilawati, H. (2018).

    Novel Synthesis of Nanoparticles from Cockle Shells via Mechanical Method

    for Cytarabine Drug Release. Journal of Computational and Theoretical

    Nanoscience, 15 (4), 1128-1136.

    Gilman, A. (1946). Therapeutic applications of chemical warfare agents. Paper

    presented at the Federation proceedings.

    Gökbuget, N., Hartog, C.-M., Bassan, R., Derigs, H.-G., Dombret, H., Greil, R.,

    Hernández-Rivas, J.-M., Huguet, F., Intermesoli, T., and Jourdan, E. (2011).

    Liposomal cytarabine is effective and tolerable in the treatment of central

    nervous system relapse of acute lymphoblastic leukemia and very aggressive

    lymphoma. Haematologica, 96 (2), 238-244.

    Goldin, A., Venditti, J. M., Kline, I., and Mantel, N. (1960). Evaluation of

    antileukemic agents employing advanced leukemia L1210 in mice II. Cancer

    Res, 20 (5 Part 2), 382-448.

    Goodman, L. S., Wintrobe, M. M., Dameshek, W., Goodman, M., Gilman, A., and

    McLennan, M. T. (1946). Nitrogen mustard therapy. Jama, 132 (3), 126-132.

    Goodman, M., Yoshizawa, C., and Kolonel, L. (1989). Incidence trends and ethnic

    patterns for childhood leukaemia in Hawaii: 1960-1984. British journal of

    cancer, 60 (1), 93-97.

    Grant, S. (1997). Ara-C: cellular and molecular pharmacology. Advances in cancer

    research, 72, 197-233.

    Grasby, S. E. (2003). Naturally precipitating vaterite (μ-CaCO 3) spheres: unusual

    carbonates formed in an extreme environment. Geochimica et Cosmochimica

    Acta, 67 (9), 1659-1666.

    Greish, K., Sawa, T., Fang, J., Akaike, T., and Maeda, H. (2004). SMA–doxorubicin,

    a new polymeric micellar drug for effective targeting to solid tumours. Journal

    of Controlled Release, 97 (2), 219-230.

    Guo, D., Zhu, L., Huang, Z., Zhou, H., Ge, Y., Ma, W., Wu, J., Zhang, X., Zhou, X.,

    and Zhang, Y. (2013). Anti-leukemia activity of PVP-coated silver

    nanoparticles via generation of reactive oxygen species and release of silver

    ions. Biomaterials, 34 (32), 7884-7894.

    Guo, X., and Szoka, F. C. (2003). Chemical approaches to triggerable lipid vesicles

    for drug and gene delivery. Accounts of chemical research, 36 (5), 335-341.

    Guo, Y., Zhang, J., Jiang, L., Shi, X., Yang, L., Fang, Q., Fang, H., Wang, K., and

    Jiang, K. (2012). Facile one-pot preparation of calcite mesoporous carrier for

    sustained and targeted drug release for cancer cells. Chemical

    Communications, 48 (86), 10636-10638.

  • © CO

    PYRI

    GHT U

    PM

    128

    Guzman, M. L., and Allan, J. N. (2014). Concise review: leukemia stem cells in

    personalized medicine. Stem cells, 32 (4), 844-851.

    Hamada, A., Kawaguchi, T., and Nakano, M. (2002). Clinical pharmacokinetics of

    cytarabine formulations. Clinical pharmacokinetics, 41 (10), 705-718.

    Hammadi, I. N., Abba, Y., Hezmee, M. N. M., Razak, I. S. A., Jaji, A. Z., Isa, T.,

    Mahmood, S. K., and Zakaria, M. Z. A. B. (2017a). Formulation of a Sustained

    Release Docetaxel Loaded Cockle Shell-Derived Calcium Carbonate

    Nanoparticles against Breast Cancer. Pharmaceutical Research, 34 (6), 1193-

    1203.

    Hammadi, N. I., Abba, Y., Hezmee, M. N. M., Razak, I. S. A., Kura, A. U., and

    Zakaria, Z. A. B. (2017c). Evaluation of in vitro efficacy of docetaxel-loaded

    calcium carbonate aragonite nanoparticles (DTX-CaCO3NP) on 4T1 mouse

    breast cancer cell line. In Vitro Cellular & Developmental Biology-Animal, 53

    (10), 896-907.

    Hartwell, L. H., and Kastan, M. B. (1994). Cell cycle control and cancer. Science, 266

    (5192), 1821-1828.

    He, X., Liu, T., Chen, Y., Cheng, D., Li, X., Xiao, Y., and Feng, Y. (2008). Calcium

    carbonate nanoparticle delivering vascular endothelial growth factor-C siRNA

    effectively inhibits lymphangiogenesis and growth of gastric cancer in vivo.

    Cancer gene therapy, 15 (3), 193.

    Herzig, R. H., Hines, J., Herzig, G., Wolff, S., Cassileth, P., Lazarus, H., Adelstein,

    D., Brown, R., Coccia, P., and Strandjord, S. (1987). Cerebellar toxicity with

    high-dose cytosine arabinoside. Journal of Clinical Oncology, 5 (6), 927-932.

    Herzig, R. H., Lazarus, H. M., Wolff, S. N., Phillips, G. L., and Herzig, G. P. (1985).

    High-dose cytosine arabinoside therapy with and without anthracycline

    antibiotics for remission reinduction of acute nonlymphoblastic leukemia.

    Journal of Clinical Oncology, 3 (7), 992-997.

    Herzig, R. H., Wolff, S. N., Lazarus, H. M., Phillips, G. L., Karanes, C., and Herzig,

    G. P. (1983). High-dose cytosine arabinoside therapy for refractory leukemia.

    Blood, 62 (2), 361-369.

    Hesselton, R. M., Greiner, D. L., Mordes, J. P., Rajan, T. V., Sullivan, J. L., and Shultz,

    L. D. (1995). High levels of human peripheral blood mononuclear cell

    engraftment and enhanced susceptibility to human immunodeficiency virus

    type 1 infection in NOD/LtSz-scid/scid mice. Journal of Infectious Diseases,

    172 (4), 974-982.

    Hobson, D. W. (2009). Commercialization of nanotechnology. Wiley Interdisciplinary

    Reviews: Nanomedicine and Nanobiotechnology, 1 (2), 189-202.

  • © CO

    PYRI

    GHT U

    PM

    129

    Hoelzer, D., Gökbuget, N., Ottmann, O., Pui, C.-H., Relling, M. V., Appelbaum, F.

    R., van Dongen, J. J., and Szczepański, T. (2002). Acute lymphoblastic

    leukemia. ASH Education Program Book, 2002 (1), 162-192.

    Honary, S., and Zahir, F. (2013). Effect of zeta potential on the properties of nano-

    drug delivery systems-a review (Part 2). Tropical Journal of Pharmaceutical

    Research, 12 (2), 265-273.

    Huang, J., and Heise, A. (2013). Stimuli responsive synthetic polypeptides derived

    from N-carboxyanhydride (NCA) polymerisation. Chemical Society Reviews,

    42 (17), 7373-7390.

    Huang, S., Chen, J. C., Hsu, C. W., and Chang, W. H. (2009). Effects of nano calcium

    carbonate and nano calcium citrate on toxicity in ICR mice and on bone

    mineral density in an ovariectomized mice model. Nanotechnology, 20 (37),

    375102.

    Hubert-Habari, M., and Cohen, S. S. (1962). The toxicity of 9-β-D-

    arabinofuranosyladenine to purine-requiring Escherichia coli. Biochimica et

    biophysica acta, 59 (2), 468-471.

    Ibrahim, A. R. A. E. B. (2012). Prognostic factors in Chronic Myeloid Leukaemia

    therapy.

    Ikoma, T., Tonegawa, T., Watanaba, H., Chen, G., Tanaka, J., and Mizushima, Y.

    (2007). Drug-supported microparticles of calcium carbonate nanocrystals and

    its covering with hydroxyapatite. Journal of nanoscience and nanotechnology,

    7 (3), 822-827.

    Irons, R. D., Chen, Y., Wang, X., Ryder, J., and Kerzic, P. J. (2013). Acute myeloid

    leukemia following exposure to benzene more closely resembles de novo than

    therapy related‐disease. Genes, Chromosomes and Cancer, 52 (10), 887-894.

    Islam, K. N., Bakar, M. Z. B. A., Noordin, M. M., Hussein, M. Z. B., Rahman, N. S.

    B. A., and Ali, M. E. (2011). Characterisation of calcium carbonate and its

    polymorphs from cockle shells (Anadara granosa). Powder technology, 213

    (1-3), 188-191.

    Islam, K. N., Zuki, A., Ali, M., Hussein, M. Z. B., Noordin, M., Loqman, M., Wahid,

    H., Hakim, M., and Hamid, S. B. A. (2012). Facile synthesis of calcium

    carbonate nanoparticles from cockle shells. Journal of Nanomaterials, 2012,

    2.

    Islan, G. A., Cacicedo, M. L., Bosio, V. E., and Castro, G. R. (2015). Development

    and characterization of new enzymatic modified hybrid calcium carbonate

    microparticles to obtain nano-architectured surfaces for enhanced drug

    loading. Journal of colloid and interface science, 439, 76-87.

  • © CO

    PYRI

    GHT U

    PM

    130

    Jain, R. K., and Stylianopoulos, T. (2010). Delivering nanomedicine to solid tumors.

    Nature reviews Clinical oncology, 7 (11), 653-664.

    Jain, S., Sharma, R. K., and Vyas, S. (2006). Chitosan nanoparticles encapsulated

    vesicular systems for oral immunization: preparation, in‐vitro and in‐vivo characterization. Journal of pharmacy and pharmacology, 58 (3), 303-310.

    Jaji, A. Z., Bakar, M. Z. B. A., Mahmud, R., Loqman, M. Y., Hezmee, M. N. M., Isa,

    T., Wenliang, F., and Hammadi, N. I. (2017). Synthesis, characterization, and

    cytocompatibility of potential cockle shell aragonite nanocrystals for

    osteoporosis therapy and hormonal delivery. Nanotechnology, science and

    applications, 10, 23.

    Janes, K. A., Fresneau, M. P., Marazuela, A., Fabra, A., and Alonso, M. a. J. (2001).

    Chitosan nanoparticles as delivery systems for doxorubicin. Journal of

    Controlled Release, 73 (2-3), 255-267.

    Jemal, A., Siegel, R., Xu, J., and Ward, E. (2010). Cancer statistics, 2010. CA: A

    Cancer Journal for Clinicians, 60 (5), 277-300.

    Jemal, A., Thomas, A., Murray, T., and Thun, M. (2002). Cancer statistics, 2002. CA:

    A Cancer Journal for Clinicians, 52 (1), 23-47.

    Ji, Q., and Qiu, L. (2016). Mechanism study of PEGylated polyester and β-

    cyclodextrin integrated micelles on drug resistance reversal in MRP1-

    overexpressed HL60/ADR cells. Colloids and Surfaces B: Biointerfaces, 144,

    203-213.

    Jiang, W. G., Sanders, A. J., Katoh, M., Ungefroren, H., Gieseler, F., Prince, M.,

    Thompson, S., Zollo, M., Spano, D., and Dhawan, P. (2015). Tissue invasion

    and metastasis: Molecular, biological and clinical perspectives. Paper

    presented at the Seminars in cancer biology.

    Jin, J., Jiang, D., Mai, W., Meng, H., Qian, W., Tong, H., Huang, J., Mao, L., Tong,

    Y., and Wang, L. (2006). Homoharringtonine in combination with cytarabine

    and aclarubicin resulted in high complete remission rate after the first induction

    therapy in patients with de novo acute myeloid leukemia. Leukemia, 20 (8),

    1361-1367.

    Jing, L., Liang, X., Li, X., Yang, Y., and Dai, Z. (2013). Covalent attachment of Mn-

    porphyrin onto doxorubicin-loaded poly (lactic acid) nanoparticles for

    potential magnetic resonance imaging and pH-sensitive drug delivery. Acta

    biomaterialia, 9 (12), 9434-9441.

    Ju, X.-J., Liu, L., Xie, R., Niu, C. H., and Chu, L.-Y. (2009a). Dual thermo-responsive

    and ion-recognizable monodisperse microspheres. Polymer, 50 (3), 922-929.

  • © CO

    PYRI

    GHT U

    PM

    131

    Ju, X.-J., Xie, R., Yang, L., and Chu, L.-Y. (2009b). Biodegradable

    ‘intelligent’materials in response to chemical stimuli for biomedical

    applications. Expert opinion on therapeutic patents, 19 (5), 683-696.

    Ju, X.-J., Xie, R., Yang, L., and Chu, L.-Y. (2009c). Biodegradable

    ‘intelligent’materials in response to physical stimuli for biomedical

    applications. Expert opinion on therapeutic patents, 19 (4), 493-507.

    Kamba, A. S., Ismail, M., Ibrahim, T. A. T., and Zakaria, Z. A. B. (2013a). Synthesis

    and characterisation of calcium carbonate aragonite nanocrystals from cockle

    shell powder (Anadara granosa). Journal of Nanomaterials, 2013, 5.

    Kamba, A. S., Ismail, M., Ibrahim, T. A. T., and Zakaria, Z. A. B. (2014).

    Biocompatibility of bio based calcium carbonate nanocrystals aragonite

    polymorph on NIH 3T3 fibroblast cell line. African Journal of Traditional,

    Complementary and Alternative Medicines, 11 (4), 31-38.

    Kamba, S. A., Ismail, M., Hussein-Al-Ali, S. H., Ibrahim, T. A. T., and Zakaria, Z. A.

    B. (2013b). In vitro delivery and controlled release of doxorubicin for targeting

    osteosarcoma bone cancer. Molecules, 18 (9), 10580-10598.

    Karamitros, C. S., Yashchenok, A. M., Möhwald, H., Skirtach, A. G., and Konrad, M.

    (2013). Preserving catalytic activity and enhancing biochemical stability of the

    therapeutic enzyme asparaginase by biocompatible multilayered

    polyelectrolyte microcapsules. Biomacromolecules, 14 (12), 4398-4406.

    Kashi, T. S. J., Eskandarion, S., Esfandyari-Manesh, M., Marashi, S. M. A., Samadi,

    N., Fatemi, S. M., Atyabi, F., Eshraghi, S., and Dinarvand, R. (2012).

    Improved drug loading and antibacterial activity of minocycline-loaded PLGA

    nanoparticles prepared by solid/oil/water ion pairing method. International

    Journal of Nanomedicine, 7, 221.

    Khalafalla, M. M., Abdellatef, E., Daffalla, H. M., Nassrallah, A. A., Aboul-Enein, K.

    M., Lightfoot, D. A., Cocchetto, A., and El-Shemy, H. A. (2009).

    Antileukemia activity from root cultures of Vernonia amygdalina. Journal of

    Medicinal Plants Research, 3 (8), 556-562.

    Kim, J.-H., Kim, Y.-S., Park, K., Lee, S., Nam, H. Y., Min, K. H., Jo, H. G., Park, J.

    H., Choi, K., and Jeong, S. Y. (2008). Antitumor efficacy of cisplatin-loaded

    glycol chitosan nanoparticles in tumor-bearing mice. Journal of Controlled

    Release, 127 (1), 41-49.

    Kim, S. K., Foote, M. B., and Huang, L. (2013). Targeted delivery of EV peptide to

    tumor cell cytoplasm using lipid coated calcium carbonate nanoparticles.

    Cancer letters, 334 (2), 311-318.

    Knoester, P., Underberg, W., and Beijnen, J. (1993). Clinical pharmacokinetics and

    pharmacodynamics of anticancer agents in pediatric patients. Anticancer

    research, 13 (5C), 1795-1808.

  • © CO

    PYRI

    GHT U

    PM

    132

    Koo, A. N., Min, K. H., Lee, H. J., Jegal, J. H., Lee, J. W., and Lee, S. C. (2015).

    Calcium Carbonate Mineralized Nanoparticles as an Intracellular Transporter

    of Cytochrome c for Cancer Therapy. Chemistry–An Asian Journal, 10 (11),

    2380-2387.

    Korach, C. S., and Pai, R. K. (2011). Mechanical Properties of a Nanostructured Poly

    (KAMPS)/aragonite Composite Mechanics of Biological Systems and

    Materials, Volume 2 (pp. 131-136): Springer.

    Krause, D. S., and Van Etten, R. A. (2007). Right on target: eradicating leukemic stem

    cells. Trends in molecular medicine, 13 (11), 470-481.

    Lamba, J. K. (2009). Genetic factors influencing cytarabine therapy.

    Pharmacogenomics, 10 (10), 1657-1674.

    Lanzilli, G., Fuggetta, M. P., Tricarico, M., Cottarelli, A., Serafino, A., Falchetti, R.,

    Ravagnan, G., Turriziani, M., Adamo, R., and Franzese, O. (2006). Resveratrol

    down-regulates the growth and telomerase activity of breast cancer cells in

    vitro. International journal of oncology, 28 (3), 641-648.

    Lauzon, G. J., Paterson, A. R., and Belch, A. W. (1978). Formation of 1-β-D-

    arabinofuranosylcytosine diphosphate choline in neoplastic and normal cells.

    Cancer Res, 38 (6), 1730-1733.

    Leclerc, J.-M., and Momparler, R. L. (1984). Effect of the interval between exposures

    to cytarabine on its cytotoxic action on HL-60 myeloid leukemic cells. Cancer

    treatment reports, 68 (9), 1143-1148.

    Lee, H. J., Park, D. J., Choi, G. H., Yang, D.-N., Heo, J. S., and Lee, S. C. (2016). pH-

    Responsive mineralized nanoparticles as stable nanocarriers for intracellular

    nitric oxide delivery. Colloids and Surfaces B: Biointerfaces, 146, 1-8.

    LePage, G., Lin, Y.-T., Orth, R., and Gottlieb, J. (1972). 5′-Nucleotides as potential

    formulations for administering nucleoside analogs in man. Cancer Res, 32

    (11), 2441-2444.

    Levy, M., and Benita, S. (1990). Drug release from submicronized o/w emulsion: a

    new in vitro kinetic evaluation model. International journal of pharmaceutics,

    66 (1-3), 29-37.

    Li, L., Nurunnabi, M., Nafiujjaman, M., Lee, Y.-k., and Huh, K. M. (2013). GSH-

    mediated photoactivity of pheophorbide a-conjugated heparin/gold

    nanoparticle for photodynamic therapy. Journal of Controlled Release, 171

    (2), 241-250.

    Lim, G., Yahaya, H., and Lim, T. (2003). The first report of the national cancer registry

    cancer incidence in Malaysia. Kuala Lumpur: Ministry of Health, 57-58.

  • © CO

    PYRI

    GHT U

    PM

    133

    Liu, J., Huang, Y., Kumar, A., Tan, A., Jin, S., Mozhi, A., and Liang, X.-J. (2014).

    pH-sensitive nano-systems for drug delivery in cancer therapy. Biotechnology

    advances, 32 (4), 693-710.

    Liu, J., Zhao, D., He, W., Zhang, H., Li, Z., and Luan, Y. (2017a). Nanoassemblies

    from amphiphilic cytarabine prodrug for leukemia targeted therapy. Journal of

    colloid and interface science, 487, 239-249.

    Liu, Y., Zhi, X., Yang, M., Zhang, J., Lin, L., Zhao, X., Hou, W., Zhang, C., Zhang,

    Q., and Pan, F. (2017b). Tumor-triggered drug release from calcium carbonate-

    encapsulated gold nanostars for near-infrared photodynamic/photothermal

    combination antitumor therapy. Theranostics, 7 (6), 1650.

    Liu, Z., Liu, L., Ju, X.-J., Xie, R., Zhang, B., and Chu, L.-Y. (2011). K+-recognition

    capsules with squirting release mechanisms. Chemical Communications, 47

    (45), 12283-12285.

    Logue, J. S., and Morrison, D. K. (2012). Complexity in the signaling network:

    insights from the use of targeted inhibitors in cancer therapy. Genes &

    development, 26 (7), 641-650.

    Lombardo, A. (2009b). Toxicity and anti-tumor effects of cytarabine and doxorubicin

    on acute myeloid leukemia. A major qualifying project report submitted to the

    Faculty of the Worcester polytechnic Institute,England.

    Lønning, P. E. (2003). Study of suboptimum treatment response: lessons from breast

    cancer. The lancet oncology, 4 (3), 177-185.

    Loos, C., Syrovets, T., Musyanovych, A., Mailänder, V., Landfester, K., and Simmet,

    T. (2014). Amino-functionalized nanoparticles as inhibitors of mTOR and

    inducers of cell cycle arrest in leukemia cells. Biomaterials, 35 (6), 1944-1953.

    Löwenberg, B. (2013). Sense and nonsense of high-dose cytarabine for acute myeloid

    leukemia. Blood, 121 (1), 26-28.

    Lowenberg, B., Downing, J. R., and Burnett, A. (1999). Acute myeloid leukemia. N

    Engl J Med, 1999 (341), 1051-1062.

    Löwenberg, B., Pabst, T., Vellenga, E., van Putten, W., Schouten, H. C., Graux, C.,

    Ferrant, A., Sonneveld, P., Biemond, B. J., and Gratwohl, A. (2011).

    Cytarabine dose for acute myeloid leukemia. New England Journal of

    Medicine, 364 (11), 1027-1036.

    Luo, Y., Zhang, K., Wei, Q., Liu, Z., and Chen, Y. (2009). Poly (MAA-co-AN)

    hydrogels with improved mechanical properties for theophylline controlled

    delivery. Acta biomaterialia, 5 (1), 316-327.

  • © CO

    PYRI

    GHT U

    PM

    134

    Ma, P. (2012). Drug-Loaded Lipid Nanoparticles for Improved Cancer Treatment:

    Engineering, In-Vitro, and In-Vivo Evaluation, PhD thesis. The University of

    North Carolina, Chapel Hill.

    Mardis, E. R., Ding, L., Dooling, D. J., Larson, D. E., McLellan, M. D., Chen, K.,

    Koboldt, D. C., Fulton, R. S., Delehaunty, K. D., and McGrath, S. D. (2009).

    Recurring mutations found by sequencing an acute myeloid leukemia genome.

    New England Journal of Medicine, 361 (11), 1058-1066.

    Martirosyan, A., and Schneider, Y.-J. (2014). Engineered nanomaterials in food:

    implications for food safety and consumer health. International journal of

    environmental research and public health, 11 (6), 5720-5750.

    Mathers, C. D., Boschi-Pinto, C., Lopez, A. D., Murray, C. J., and Organization, W.

    H. (2001). Cancer incidence, mortality and survival by site for 14 regions of

    the world.Geneva, World Health Organization (GPE Discussion Paper No.

    13).

    Matsui, W., Huff, C. A., Wang, Q., Malehorn, M. T., Barber, J., Tanhehco, Y., Smith,

    B. D., Civin, C. I., and Jones, R. J. (2004). Characterization of clonogenic

    multiple myeloma cells. Blood, 103 (6), 2332-2336.

    McTiernan, K., and O'Connell, M. (2015). An interpretative phenomenological

    analysis exploring the lived experience of individuals dying from terminal

    cancer in Ireland. Palliative & supportive care, 13 (3), 641-651.

    Meshik, X., Xu, K., Dutta, M., and Stroscio, M. A. (2014). Optical detection of lead

    and potassium ions using a quantum-dot-based aptamer nanosensor. IEEE

    transactions on nanobioscience, 13 (2), 161-164.

    Moghadam, M. N., Kolesov, V., Vogel, A., Klok, H.-A., and Pioletti, D. P. (2014).

    Controlled release from a mechanically-stimulated thermosensitive self-

    heating composite hydrogel. Biomaterials, 35 (1), 450-455.

    Moghimi, S. M., Hunter, A. C., and Murray, J. C. (2001). Long-circulating and target-

    specific nanoparticles: theory to practice. Pharmacological reviews, 53 (2),

    283-318.

    Momparler, R. L. (1974). A model for the chemotherapy of acute leukemia with 1-β-

    D-arabinofuranosylcytosine. Cancer Res, 34 (8), 1775-1787.

    Momparler, R. L. (2013). Optimization of cytarabine (ARA-C) therapy for acute

    myeloid leukemia. Experimental hematology & oncology, 2 (1), 20.

    Moore, M. (2006). Do nanoparticles present ecotoxicological risks for the health of

    the aquatic environment? Environment international, 32 (8), 967-976.

  • © CO

    PYRI

    GHT U

    PM

    135

    Murthy, V. S., Rohini, A., Pravallika, K., Rani, A. P., and Rahaman, S. (2013).

    Development and validation of rp-hplc method for estimation of cytarabine in

    bulk and pharmacutical dosage forms. International Journal of

    Pharmaceutical Sciences and Research, 4 (12), 4573.

    Nagayama, S., Ogawara, K.-i., Fukuoka, Y., Higaki, K., and Kimura, T. (2007). Time-

    dependent changes in opsonin amount associated on nanoparticles alter their

    hepatic uptake characteristics. International journal of pharmaceutics, 342 (1),

    215-221.

    Nair, M. S., Mony, U., Menon, D., Koyakutty, M., Sidharthan, N., Pavithran, K., Nair,

    S. V., and Menon, K. N. (2015). Development and molecular characterization

    of polymeric micro-nanofibrous scaffold of a defined 3-D niche for in vitro

    chemosensitivity analysis against acute myeloid leukemia cells. International

    Journal of Nanomedicine, 10, 3603.

    Naruphontjirakul, P., and Viravaidya-Pasuwat, K. (2011). Development of

    Doxorubicin-Core Shell O-succinyl Chitosan Graft Pluronic® 127 Copolymer

    Nanoparticles to Treat Human Cancer. International Journal of Bioscience,

    Biochemistry and Bioinformatics, 1 (2), 131.

    Neira-Carrillo, A., Yslas, E., Marini, Y. A., Vásquez-Quitral, P., Sánchez, M.,

    Riveros, A., Yanez, D., Cavallo, P., Kogan, M. J., and Acevedo, D. (2016).

    Hybrid biomaterials based on calcium carbonate and polyaniline nanoparticles

    for application in photothermal therapy. Colloids and Surfaces B:

    Biointerfaces, 145, 634-642.

    Neri, D., and Supuran, C. T. (2011). Interfering with pH regulation in tumours as a

    therapeutic strategy. Nature reviews Drug discovery, 10 (10), 767-777.

    Ng, S.-F., and Jumaat, N. (2014). Carboxymethyl cellulose wafers containing

    antimicrobials: A modern drug delivery system for wound infections.

    European Journal of Pharmaceutical Sciences, 51, 173-179.

    Nicholson, J. K., Connelly, J., Lindon, J. C., and Holmes, E. (2002). Metabonomics:

    a platform for studying drug toxicity and gene function. Nature reviews Drug

    discovery, 1 (2), 153-161.

    O'brien, S. G., Guilhot, F.